Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.769
Filter
1.
Neurosurg Rev ; 47(1): 305, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38967704

ABSTRACT

The clinical management of aneurysmal subarachnoid hemorrhage (SAH)-associated vasospasm remains a challenge in neurosurgical practice, with its prevention and treatment having a major impact on neurological outcome. While considered a mainstay, nimodipine is burdened by some non-negligible limitations that make it still a suboptimal candidate of pharmacotherapy for SAH. This narrative review aims to provide an update on the pharmacodynamics, pharmacokinetics, overall evidence, and strength of recommendation of nimodipine alternative drugs for aneurysmal SAH-associated vasospasm and delayed cerebral ischemia. A PRISMA literature search was performed in the PubMed/Medline, Web of Science, ClinicalTrials.gov, and PubChem databases using a combination of the MeSH terms "medical therapy," "management," "cerebral vasospasm," "subarachnoid hemorrhage," and "delayed cerebral ischemia." Collected articles were reviewed for typology and relevance prior to final inclusion. A total of 346 articles were initially collected. The identification, screening, eligibility, and inclusion process resulted in the selection of 59 studies. Nicardipine and cilostazol, which have longer half-lives than nimodipine, had robust evidence of efficacy and safety. Eicosapentaenoic acid, dapsone and clazosentan showed a good balance between effectiveness and favorable pharmacokinetics. Combinations between different drug classes have been studied to a very limited extent. Nicardipine, cilostazol, Rho-kinase inhibitors, and clazosentan proved their better pharmacokinetic profiles compared with nimodipine without prejudice with effective and safe neuroprotective role. However, the number of trials conducted is significantly lower than for nimodipine. Aneurysmal SAH-associated vasospasm remains an area of ongoing preclinical and clinical research where the search for new drugs or associations is critical.


Subject(s)
Brain Ischemia , Neuroprotective Agents , Nimodipine , Subarachnoid Hemorrhage , Vasospasm, Intracranial , Humans , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/drug therapy , Vasospasm, Intracranial/drug therapy , Vasospasm, Intracranial/etiology , Nimodipine/therapeutic use , Brain Ischemia/drug therapy , Neuroprotective Agents/therapeutic use , Neuroprotection/drug effects , Cilostazol/therapeutic use , Nicardipine/therapeutic use , Dioxanes/therapeutic use , Vasodilator Agents/therapeutic use , Pyrimidines/therapeutic use , Pyridines , Sulfonamides , Tetrazoles
2.
Sci Rep ; 14(1): 15153, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956206

ABSTRACT

Durian (Durio zibethinus L.) fruit pulp is a rich source of γ-glutamylcysteine (γ-EC), a direct precursor to the antioxidant glutathione (GSH). This study elucidated the in vitro neuroprotective potential of unripe durian fruit pulp extract (UDE) against H2O2-induced neurotoxicity in SH-SY5Y cells and neuroinflammation in lipopolysaccharide (LPS)-stimulated BV-2 cells. Treatments with γ-EC, GSH standards, or UDE exhibited no cytotoxicity in SH-SY5Y and BV-2 cells, except at high concentrations. A 4-h pretreatment with 100 µM γ-EC or UDE containing 100 µM γ-EC significantly increased SH-SY5Y cell viability post H2O2 induction. Moreover, a similar pretreatment reduced LPS-stimulated production of proinflammatory cytokines in BV-2 cells. The neuroprotective effect of UDE is primarily attributed to γ-EC provision and the promotion of GSH synthesis, which in turn elevates intracellular GSH levels and reduces proinflammatory cytokines. This study identifies γ-EC in UDE as a potential neuroprotective biomarker boosting intracellular GSH levels, providing insights into UDE's therapeutic potential.


Subject(s)
Fruit , Glutathione , Neuroprotective Agents , Oxidative Stress , Plant Extracts , Glutathione/metabolism , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Neuroprotective Agents/pharmacology , Humans , Fruit/chemistry , Animals , Inflammation/metabolism , Inflammation/drug therapy , Lipopolysaccharides , Neuroprotection/drug effects , Mice , Cell Survival/drug effects , Hydrogen Peroxide/metabolism , Antioxidants/pharmacology , Cell Line, Tumor , Cell Line , Cytokines/metabolism , Dipeptides/pharmacology
3.
ACS Nano ; 18(26): 16853-16866, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38896491

ABSTRACT

The precise modulation of electrical activity in specific neuronal populations is paramount for rectifying abnormal neurological functions and is a critical element in the therapeutic arsenal for neurological disorders. However, achieving a balance between minimal invasiveness and robust neuroprotection poses a considerable challenge. Herein, we present a nanoneuromodulation strategy integrating neuroprotective features to effectively address epilepsy with minimal invasiveness and enable wireless functionality. Strategically engineered nanotransducer, adorned with platinum (Pt) decoration with titanium disulfide (TiS2) (TiS2/Pt), enables precise modulation of neuronal electrical activity in vitro and in vivo, ensuring exceptional temporal fidelity under millisecond-precision near-infrared (NIR) light pulses irradiation. Concurrently, TiS2/Pt showcase a pronounced enhancement in enzyme-mimicking activity, offering a robust defense against oxidative neurological injury in vitro. Nanotransducer-enabled wireless neuromodulation with biocatalytic neuroprotective capacity is highly effective in alleviating epileptic high-frequency neural activity and diminishing oxidative stress levels, thereby restoring redox equilibrium. This integrated therapeutic approach reduces the severity of epilepsy, demonstrating minimal invasiveness and obviating the requirements for genetic manipulation and optical fiber implantation, while providing an alternative avenue for neurological disorder treatment.


Subject(s)
Epilepsy , Epilepsy/therapy , Animals , Titanium/chemistry , Titanium/pharmacology , Platinum/chemistry , Platinum/pharmacology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Neuroprotection/drug effects , Mice , Disulfides/chemistry , Disulfides/pharmacology , Oxidative Stress/drug effects , Humans , Neurons/drug effects , Neurons/metabolism , Infrared Rays , Rats
4.
CNS Neurosci Ther ; 30(6): e14795, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38867401

ABSTRACT

AIM: Recent studies have extensively investigated hypothermia as a therapeutic approach for mitigating neural damage. Despite this, bibliometric analyses specifically focusing on this area remain scarce. Consequently, this study aims to comprehensively outline the historical framework of research and to pinpoint future research directions and trends. METHODS: Articles spanning from 2003 to 2023, relevant to both "neuroprotection" and "hypothermia", were sourced from the Web of Science Core Collection. The CiteSpace software facilitated a comprehensive evaluation and analysis of these publications. This analysis included examining the annual productivity, collaboration among nations, institutions, and authors, as well as the network of co-cited references, authors and journals, and the co-occurrence of keywords, and their respective clusters and trends, all of which were visualized. RESULTS: This study included 2103 articles on the neuroprotection effects of hypothermia, noting a consistent increase in publications since 1992. The United States, the University of California System, and Ji Xunming emerged as the most productive nation, institution, and author, respectively. Analysis of the top 10 co-cited publications revealed that seven articles focused on the effects of hypothermia in infants with hypoxic-ischemic encephalopathy, while three studies addressed cardiac arrest. Shankaran S and the journal Stroke were the most frequently co-cited author and journal, respectively. Keyword cluster analysis identified ischemic stroke as the primary focus of hypothermia therapy historically, with cardiac arrest and neonatal hypoxic-ischemic encephalopathy emerging as current research foci. CONCLUSIONS: Recent studies on the neuroprotective effects of hypothermia in cardiac arrest and neonatal hypoxic-ischemic encephalopathy suggest that hypothermia may mitigate neural damage associated with these conditions. However, the application of hypothermia in the treatment of ischemic stroke remains confined to animal models and in vitro studies, with a notable absence of evidence from multicenter randomized controlled trials (RCTs). Further research is required to address this gap.


Subject(s)
Bibliometrics , Hypothermia, Induced , Neuroprotection , Hypothermia, Induced/trends , Hypothermia, Induced/methods , Humans , Neuroprotection/physiology , Animals , Hypoxia-Ischemia, Brain/therapy
5.
Int J Mol Sci ; 25(11)2024 May 28.
Article in English | MEDLINE | ID: mdl-38892094

ABSTRACT

Alzheimer's Disease (AD), a prevalent neurodegenerative disorder, is the primary cause of dementia. Despite significant advancements in neuroscience, a definitive cure or treatment for this debilitating disease remains elusive. A notable characteristic of AD is oxidative stress, which has been identified as a potential therapeutic target. Polyphenols, secondary metabolites of plant origin, have attracted attention due to their potent antioxidant properties. Epidemiological studies suggest a correlation between the consumption of polyphenol-rich foods and the prevention of chronic diseases, including neurodegenerative disorders, which underscores the potential of polyphenols as a therapeutic strategy in AD management. Hence, this comprehensive review focuses on the diverse roles of polyphenols in AD, with a particular emphasis on neuroprotective potential. Scopus, ScienceDirect, and Google Scholar were used as leading databases for study selection, from 2018 to late March 2024. Analytical chemistry serves as a crucial tool for characterizing polyphenols, with a nuanced exploration of their extraction methods from various sources, often employing chemometric techniques for a holistic interpretation of the advances in this field. Moreover, this review examines current in vitro and in vivo research, aiming to enhance the understanding of polyphenols' role in AD, and providing valuable insights for forthcoming approaches in this context.


Subject(s)
Alzheimer Disease , Neuroprotective Agents , Polyphenols , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Polyphenols/therapeutic use , Polyphenols/chemistry , Polyphenols/pharmacology , Humans , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Animals , Oxidative Stress/drug effects , Antioxidants/therapeutic use , Antioxidants/pharmacology , Neuroprotection/drug effects
6.
Biomed Pharmacother ; 176: 116830, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38824833

ABSTRACT

Growth factor-derived peptides are bioactive molecules that play a crucial role in various physiological processes within the human body. Over the years, extensive research has revealed their diverse applications, ranging from antimicrobial properties to their potential in neuroprotection and treating various diseases. These peptides exhibit innate immune responses and have been found to possess potent antimicrobial properties against a wide range of pathogens. Growth factor-derived peptides have demonstrated the ability to promote neuronal survival, prevent cell death, and stimulate neural regeneration. As a result, they hold immense promise in the treatment of various neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and multiple sclerosis, as well as in the management of traumatic brain injuries. Moreover, growth factor-derived peptides have shown potential for supporting tissue repair and wound healing processes. By enhancing cell proliferation and migration, these peptides contribute to the regeneration of damaged tissues and promote a more efficient healing response. The applications of growth factor-derived peptides extend beyond their therapeutic potential in health; they also have a role in various disease conditions. For example, researchers have explored their influence on cancer cells, where some peptides have demonstrated anti-cancer properties, inhibiting tumor growth and promoting apoptosis in cancer cells. Additionally, their immunomodulatory properties have been investigated for potential applications in autoimmune disorders. Despite the immense promise shown by growth factor-derived peptides, some challenges need to be addressed. Nevertheless, ongoing research and advancements in biotechnology offer promising avenues to overcome these obstacles. The review summarizes the foundational biology of growth factors and the intricate signaling pathways in various physiological processes as well as diseases such as cancer, neurodegenerative disorders, cardiovascular ailments, and metabolic syndromes.


Subject(s)
Intercellular Signaling Peptides and Proteins , Neuroprotective Agents , Humans , Animals , Intercellular Signaling Peptides and Proteins/pharmacology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Anti-Infective Agents/pharmacology , Neurodegenerative Diseases/drug therapy , Neuroprotection/drug effects , Peptides/pharmacology
7.
Biomater Sci ; 12(14): 3610-3621, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38842122

ABSTRACT

Oxidative stress (OS) plays an important role in the emergence and prevention of neurodegenerative diseases, such as Alzheimer's disease (AD). Excess reactive oxygen species (ROS) accumulated in a neuronal cell can lead to OS, producing cell injury and death. Seeking nanoantioxidants against AD-related oxidative stress has attracted a lot of attention, especially those potential antioxidant agents derived from natural polyphenols. However, the transformation of abundant plant polyphenols to antioxidative biomaterials against OS is still challenging. In this work, we report a new method to transform amorphous tannic acid (TA) into tailorable shaped ellagic acid (EA) crystalline particles without using an organic solvent. EA crystalline particles were generated from TA, which underwent a chemical transformation, in situ metal phenolic coordination and acid-induced assembly process, and the size and shape could be controlled by varying the amount of acid. As-prepared EA crystalline particles showed excellent stability in water and lysosomal mimicking fluid and possess unique fluorescence properties and a strong response in mass spectrometry, which is beneficial for their imaging analysis in cells and tissues. More importantly, EA particles have shown significant H2O2-related ROS scavenging ability, a high cellular uptake capacity, an excellent neuroprotective effect in PC12 cells, a high drug loading capacity and BBB permeability to enter the brain. Our study suggested that the EA crystalline particles show great potential for OS-mediated AD treatment.


Subject(s)
Ellagic Acid , Neuroprotective Agents , Oxidative Stress , Reactive Oxygen Species , Tannins , Ellagic Acid/pharmacology , Ellagic Acid/chemistry , Tannins/pharmacology , Tannins/chemistry , Oxidative Stress/drug effects , PC12 Cells , Animals , Rats , Reactive Oxygen Species/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Neuroprotective Agents/chemical synthesis , Antioxidants/pharmacology , Antioxidants/chemistry , Antioxidants/chemical synthesis , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/chemistry , Neuroprotection/drug effects , Green Chemistry Technology , Polyphenols
8.
Neurochem Res ; 49(8): 2249-2270, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38837092

ABSTRACT

Morphine (Mor) has exhibited efficacy in safeguarding neurons against ischemic injuries by simulating ischemic/hypoxic preconditioning (I/HPC). Concurrently, autophagy plays a pivotal role in neuronal survival during IPC against ischemic stroke. However, the involvement of autophagy in Mor-induced neuroprotection and the potential mechanisms remain elusive. Our experiments further confirmed the effect of Mor in cellular and animal models of ischemic stroke and explored its potential mechanism. The findings revealed that Mor enhanced cell viability in a dose-dependent manner by augmenting autophagy levels and autophagic flux in neurons subjected to oxygen-glucose deprivation/reoxygenation (OGD/R). Pretreatment of Mor improved neurological outcome and reduced infarct size in mice with middle cerebral artery occlusion/reperfusion (MCAO/R) at 1, 7 and 14 days. Moreover, the use of autophagy inhibitors nullified the protective effects of Mor, leading to reactive oxygen species (ROS) accumulation, increased loss of mitochondrial membrane potential (MMP) and neuronal apoptosis in OGD/R neurons. Results further demonstrated that Mor-induced autophagy activation was regulated by mTOR-independent activation of the c-Jun NH2- terminal kinase (JNK)1/2 Pathway, both in vitro and in vivo. Overall, these findings suggested Mor-induced neuroprotection by activating autophagy, which were regulated by JNK1/2 pathway in ischemic stroke.


Subject(s)
Autophagy , Ischemic Stroke , Morphine , Neuroprotective Agents , TOR Serine-Threonine Kinases , Animals , Autophagy/drug effects , TOR Serine-Threonine Kinases/metabolism , Male , Ischemic Stroke/drug therapy , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Morphine/pharmacology , Morphine/therapeutic use , Mice , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Neurons/drug effects , Neurons/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Neuroprotection/drug effects , Neuroprotection/physiology , Cell Survival/drug effects , Cell Survival/physiology , Reactive Oxygen Species/metabolism
9.
Crit Care Nurs Clin North Am ; 36(2): 167-184, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38705686

ABSTRACT

Caring for extremely preterm infants in the neonatal intensive care unit (NICU) is a multidisciplinary team effort. A clear understanding of roles for each member of the delivery team, anticipation of challenges, and standardized checklists support improved outcomes for this population. Physicians and nursing leaders are responsible for being role models and holding staff accountable for creating a unit culture of Neuroprotective Infant and Family-Centered Developmental Care. It is essential for parents to be included as part of the care team and babies to be acknowledged for their efforts in coping with the developmentally unexpected NICU environment.


Subject(s)
Intensive Care Units, Neonatal , Humans , Intensive Care Units, Neonatal/organization & administration , Infant, Newborn , Infant, Extremely Premature , Patient Care Team , Parents/psychology , Parents/education , Neuroprotection , Child Development/physiology , Intensive Care, Neonatal/organization & administration
10.
Redox Biol ; 73: 103169, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38692093

ABSTRACT

BACKGROUND: Inflammation and subsequent mitochondrial dysfunction and cell death worsen outcomes after revascularization in ischemic stroke. Receptor-interacting protein kinase 1 (RIPK1) activated dynamin-related protein 1 (DRP1) in a NLRPyrin domain containing 3 (NLRP3) inflammasome-dependent fashion and Hypoxia-Inducible Factor (HIF)-1α play key roles in the process. This study determined how phenothiazine drugs (chlorpromazine and promethazine (C + P)) with the hypothermic and normothermic modality impacts the RIPK1/RIPK3-DRP1 and HIF-1α pathways in providing neuroprotection. METHODS: A total of 150 adult male Sprague-Dawley rats were subjected to 2 h middle cerebral artery occlusion (MCAO) followed by 24 h reperfusion. 8 mg/kg of C + P was administered at onset of reperfusion. Infarct volumes, mRNA and protein expressions of HIF-1α, RIPK1, RIPK3, DRP-1, NLRP3-inflammation and cytochrome c-apoptosis were assessed. Apoptotic cell death, infiltration of neutrophils and macrophages, and mitochondrial function were evaluated. Interaction between RIPK1/RIPK3 and HIF-1α/NLRP3 were determined. In SH-SY5Y cells subjected to oxygen/glucose deprivation (OGD), the normothermic effect of C + P on inflammation and apoptosis were examined. RESULTS: C + P significantly reduced infarct volumes, mitochondrial dysfunction (ATP and ROS concentration, citrate synthase and ATPase activity), inflammation and apoptosis with and without induced hypothermia. Overexpression of RIPK1, RIPK3, DRP-1, NLRP3-inflammasome and cytochrome c-apoptosis were all significantly reduced by C + P at 33 °C and the RIPK1 inhibitor (Nec1s), suggesting hypothermic effect of C + P via RIPK1/RIPK3-DRP1pathway. When body temperature was maintained at 37 °C, C + P and HIF-1α inhibitor (YC-1) reduced HIF-1α expression, leading to reduction in mitochondrial dysfunction, NLRP3 inflammasome and cytochrome c-apoptosis, as well as the interaction of HIF-1α and NLRP3. These were also evidenced in vitro, indicating a normothermic effect of C + P via HIF-1α. CONCLUSION: Hypothermic and normothermic neuroprotection of C + P involve different pathways. The normothermic effect was mediated by HIF-1α, while hypothermic effect was via RIPK1/RIPK3-DRP1 signaling. This provides a theoretical basis for future precise exploration of hypothermic and normothermic neuroprotection.


Subject(s)
Dynamins , Hypoxia-Inducible Factor 1, alpha Subunit , Inflammasomes , Ischemic Stroke , NLR Family, Pyrin Domain-Containing 3 Protein , Receptor-Interacting Protein Serine-Threonine Kinases , Signal Transduction , Animals , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Rats , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Ischemic Stroke/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Signal Transduction/drug effects , Inflammasomes/metabolism , Dynamins/metabolism , Dynamins/genetics , Rats, Sprague-Dawley , Phenothiazines/pharmacology , Inflammation/metabolism , Inflammation/pathology , Neuroprotection , Humans , Disease Models, Animal , Hypothermia, Induced
11.
JAMA Neurol ; 81(6): 603-610, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38709502

ABSTRACT

Importance: Out-of-hospital cardiac arrest survival rates have markedly risen in the last decades, but neurological outcome only improved marginally. Despite research on more than 20 neuroprotective strategies involving patients in comas after cardiac arrest, none have demonstrated unequivocal evidence of efficacy; however, treatment with acyl-ghrelin has shown improved functional and histological brain recovery in experimental models of cardiac arrest and was safe in a wide variety of human study populations. Objective: To determine safety and potential efficacy of intravenous acyl-ghrelin to improve neurological outcome in patients in a coma after cardiac arrest. Design, Setting, and Participants: A phase 2, double-blind, placebo-controlled, multicenter, randomized clinical trial, Ghrelin Treatment of Comatose Patients After Cardiac Arrest: A Clinical Trial to Promote Cerebral Recovery (GRECO), was conducted between January 18, 2019, and October 17, 2022. Adult patients 18 years or older who were in a comatose state after cardiac arrest were assessed for eligibility; patients were from 3 intensive care units in the Netherlands. Expected death within 48 hours or unfeasibility of treatment initiation within 12 hours were exclusion criteria. Interventions: Patients were randomized to receive intravenous acyl-ghrelin, 600 µg (intervention group), or placebo (control group) within 12 hours after cardiac arrest, continued for 7 days, twice daily, in addition to standard care. Main Outcomes and Measures: Primary outcome was the score on the Cerebral Performance Categories (CPC) scale at 6 months. Safety outcomes included any serious adverse events. Secondary outcomes were mortality and neuron-specific enolase (NSE) levels on days 1 and 3. Results: A total of 783 adult patients in a coma after cardiac arrest were assessed for eligibility, and 160 patients (median [IQR] age, 68 [57-75] years; 120 male [75%]) were enrolled. A total of 81 patients (51%) were assigned to the intervention group, and 79 (49%) were assigned to the control group. The common odds ratio (OR) for any CPC improvement in the intervention group was 1.78 (95% CI, 0.98-3.22; P = .06). This was consistent over all CPC categories. Mean (SD) NSE levels on day 1 after cardiac arrest were significantly lower in the intervention group (34 [6] µg/L vs 56 [13] µg/L; P = .04) and on day 3 (28 [6] µg/L vs 52 [14] µg/L; P = .08). Serious adverse events were comparable in incidence and type between the groups. Mortality was 37% (30 of 81) in the intervention group vs 51% (40 of 79) in the control group (absolute risk reduction, 14%; 95% CI, -2% to 29%; P = .08). Conclusions and Relevance: In patients in a coma after cardiac arrest, intravenous treatment with acyl-ghrelin was safe and potentially effective to improve neurological outcome. Phase 3 trials are needed for conclusive evidence. Trial Registration: Clinicaltrialsregister.eu: EUCTR2018-000005-23-NL.


Subject(s)
Coma , Ghrelin , Neuroprotective Agents , Humans , Male , Female , Middle Aged , Ghrelin/therapeutic use , Double-Blind Method , Aged , Coma/etiology , Neuroprotective Agents/therapeutic use , Neuroprotection/physiology , Heart Arrest/complications , Out-of-Hospital Cardiac Arrest/complications
12.
Int Immunopharmacol ; 136: 112309, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38810304

ABSTRACT

Autoimmune uveitis, a severe inflammatory condition of the eye, poses significant challenges due to its complex pathophysiology and the critical balance between protective and detrimental immune responses. Central to this balance are microglia, the resident immune cells of the central nervous system, whose roles in autoimmune uveitis are multifaceted and dynamic. This review article delves into the dual nature of microglial functions, oscillating between neuroprotective and neurotoxic outcomes in the context of autoimmune uveitis. Initially, we explore the fundamental aspects of microglia, including their activation states and basic functions, setting the stage for a deeper understanding of their involvement in autoimmune uveitis. The review then navigates through the intricate mechanisms by which microglia contribute to disease onset and progression, highlighting both their protective actions in immune regulation and tissue repair, and their shift towards a pro-inflammatory, neurotoxic profile. Special emphasis is placed on the detailed pathways and cellular interactions underpinning these dual roles. Additionally, the review examines the potential of microglial markers as diagnostic and prognostic indicators, offering insights into their clinical relevance. The article culminates in discussing future research directions, and the ongoing challenges in translating these findings into effective clinical applications. By providing a comprehensive overview of microglial mechanisms in autoimmune uveitis, this review underscores the critical balance of microglial activities and its implications for disease management and therapy development.


Subject(s)
Autoimmune Diseases , Microglia , Neuroprotection , Uveitis , Microglia/immunology , Humans , Uveitis/immunology , Animals , Autoimmune Diseases/immunology
13.
Int J Mol Sci ; 25(10)2024 May 08.
Article in English | MEDLINE | ID: mdl-38791160

ABSTRACT

While primarily produced in the pineal gland, melatonin's influence goes beyond its well-known role in regulating sleep, nighttime metabolism, and circadian rhythms, in the field of chronobiology. A plethora of new data demonstrates melatonin to be a very powerful molecule, being a potent ROS/RNS scavenger with anti-inflammatory, immunoregulatory, and oncostatic properties. Melatonin and its metabolites exert multiple beneficial effects in cutaneous and systemic aging. This review is focused on the neuroprotective role of melatonin during aging. Melatonin has an anti-aging capacity, retarding the rate of healthy brain aging and the development of age-related neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis, amyotrophic lateral sclerosis, etc. Melatonin, as well as its metabolites, N1-acetyl-N2-formyl-5-methoxykynuramine (AFMK) and N1-acetyl-5-methoxykynuramine (AMK), can reduce oxidative brain damage by shielding mitochondria from dysfunction during the aging process. Melatonin could also be implicated in the treatment of neurodegenerative conditions, by modifying their characteristic low-grade neuroinflammation. It can either prevent the initiation of inflammatory responses or attenuate the ongoing inflammation. Drawing on the current knowledge, this review discusses the potential benefits of melatonin supplementation in preventing and managing cognitive impairment and neurodegenerative diseases.


Subject(s)
Aging , Brain , Melatonin , Neurodegenerative Diseases , Neuroprotection , Neuroprotective Agents , Melatonin/metabolism , Melatonin/pharmacology , Melatonin/therapeutic use , Humans , Brain/metabolism , Brain/drug effects , Aging/metabolism , Aging/drug effects , Animals , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/drug therapy , Neuroprotection/drug effects , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Kynuramine/metabolism , Kynuramine/analogs & derivatives
14.
J Nanobiotechnology ; 22(1): 251, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750597

ABSTRACT

BACKGROUND: Hypothermia is a promising therapy for traumatic brain injury (TBI) in the clinic. However, the neuroprotective outcomes of hypothermia-treated TBI patients in clinical studies are inconsistent due to several severe side effects. Here, an injectable refrigerated hydrogel was designed to deliver 3-iodothyronamine (T1AM) to achieve a longer period of local hypothermia for TBI treatment. Hydrogel has four advantages: (1) It can be injected into injured sites after TBI, where it forms a hydrogel and avoids the side effects of whole-body cooling. (2) Hydrogels can biodegrade and be used for controlled drug release. (3) Released T1AM can induce hypothermia. (4) This hydrogel has increased medical value given its simple operation and ability to achieve timely treatment. METHODS: Pol/T hydrogels were prepared by a low-temperature mixing method and characterized. The effect of the Pol/T hydrogel on traumatic brain injury in mice was studied. The degradation of the hydrogel at the body level was observed with a small animal imager. Brain temperature and body temperature were measured by brain thermometer and body thermometer, respectively. The apoptosis of peripheral nerve cells was detected by immunohistochemical staining. The protective effect of the hydrogels on the blood-brain barrier (BBB) after TBI was evaluated by the Evans blue penetration test. The protective effect of hydrogel on brain edema after injury in mice was detected by Magnetic resonance (MR) in small animals. The enzyme linked immunosorbent assay (ELISA) method was used to measure the levels of inflammatory factors. The effects of behavioral tests on the learning ability and exercise ability of mice after injury were evaluated. RESULTS: This hydrogel was able to cool the brain to hypothermia for 12 h while maintaining body temperature within the normal range after TBI in mice. More importantly, hypothermia induced by this hydrogel leads to the maintenance of BBB integrity, the prevention of cell death, the reduction of the inflammatory response and brain edema, and the promotion of functional recovery after TBI in mice. This cooling method could be developed as a new approach for hypothermia treatment in TBI patients. CONCLUSION: Our study showed that injectable and biodegradable frozen Pol/T hydrogels to induce local hypothermia in TBI mice can be used for the treatment of traumatic brain injury.


Subject(s)
Blood-Brain Barrier , Brain Injuries, Traumatic , Hydrogels , Hypothermia, Induced , Animals , Brain Injuries, Traumatic/therapy , Brain Injuries, Traumatic/drug therapy , Mice , Hydrogels/chemistry , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Male , Hypothermia, Induced/methods , Neuroprotection/drug effects , Brain/pathology , Disease Models, Animal , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Body Temperature , Mice, Inbred C57BL
15.
Int J Mol Sci ; 25(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38791487

ABSTRACT

Hypoxic-ischemic encephalopathy (HIE) is a major cause of newborn brain damage stemming from a lack of oxygenated blood flow in the neonatal period. Twenty-five to fifty percent of asphyxiated infants who develop HIE die in the neonatal period, and about sixty percent of survivors develop long-term neurological disabilities. From the first minutes to months after the injury, a cascade of events occurs, leading to blood-brain barrier (BBB) opening, neuronal death and inflammation. To date, the only approach proposed in some cases is therapeutic hypothermia (TH). Unfortunately, TH is only partially protective and is not applicable to all neonates. This review synthesizes current knowledge on the basic molecular mechanisms of brain damage in hypoxia-ischemia (HI) and on the different therapeutic strategies in HI that have been used and explores a major limitation of unsuccessful therapeutic approaches.


Subject(s)
Hypoxia-Ischemia, Brain , Neuroprotection , Animals , Humans , Infant, Newborn , Blood-Brain Barrier/metabolism , Hypothermia, Induced/methods , Hypoxia-Ischemia, Brain/therapy , Hypoxia-Ischemia, Brain/metabolism , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Animals, Newborn
16.
Environ Pollut ; 352: 124035, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38670424

ABSTRACT

The exact mechanisms underlying the initiation and exacerbation of Parkinson's disease (PD) by paraquat remain unclear. We have revealed that exosomes mediate neurotoxicity induced by low dose paraquat exposure by transmitting intercellular signaling. Exposure to 40 µM paraquat promoted exosome release from mouse microglia cells (BV2) in vitro. Paraquat exposure at 100 µM caused degeneration of mouse dopaminergic MN9D cells and inhibited microglia exosome uptake by fluorescently labeling exosomes. We established an incubation model for exosomes and dopaminergic neuron cells under PQ treatment. The results indicated that microglial exosomes alleviated degeneration, increasing proliferation and PD-related protein expression of dopaminergic neurons; however, paraquat reversed this effect. Then, through exosome high-throughput sequencing and qRT-PCR experiments, miR-92a-3p and miR-24-3p were observed to transfer from exosomes to dopaminergic neurons, inhibited by paraquat. The specificity of miR-92a-3p and miR-24-3p was verified in PD patients exosomes, indicating the potential diagnostic value of the exosomal miRNAs in paraquat-induced PD. These results suggest glia-neuron communication in paraquat-induced neurodegeneration and may identify stable paraquat-mediated PD biomarkers, offering clues for early recognition and prevention of pesticide-induced degenerative diseases.


Subject(s)
Biomarkers , Dopaminergic Neurons , Exosomes , MicroRNAs , Microglia , Paraquat , Parkinson Disease , Paraquat/toxicity , Exosomes/metabolism , Animals , Microglia/drug effects , Microglia/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Dopaminergic Neurons/drug effects , Biomarkers/metabolism , Neuroprotection/drug effects , Humans , Cell Line
17.
ACS Chem Neurosci ; 15(11): 2223-2232, 2024 06 05.
Article in English | MEDLINE | ID: mdl-38634698

ABSTRACT

Neuronal death resulting from ischemic stroke is the primary cause of adult mortality and disability, and effective neuroprotective agents for poststroke intervention are still lacking. Remote ischemic postconditioning (RIPostC) has demonstrated significant protective effects against ischemia in various organs; however, the specific mechanisms are not fully understood. This study investigated the potential neuroprotective mechanisms of RIPostC in the context of ischemic stroke. Using a rat model of middle cerebral artery occlusion, we found that RIPostC mitigated neurological damage, improved movement in the open-field test, and protected against neuronal apoptosis. In terms of energy metabolism, RIPostC enhanced ATP levels, suppressed lactate content, and increased the production of ketone bodies (KBs). In the ferroptosis assay, RIPostC protected against lipoperoxidation, reversed the reduction of glutathione peroxidase 4 (GPX4), and mitigated the excessive expression of long-chain acyl-CoA synthetase family member 4 (ACSL4). In oxygen-glucose deprivation/reoxygenation-treated HT22 cells, KBs maintained GPX4 levels, suppressed ACSL4 expression, and preserved the mitochondrial cristae number. However, the effect of KBs on the expression of GPX4, ACSL4, and the number of mitochondrial cristae was blocked by erastin. Moreover, both RIPostC and KBs reduced total iron and ferrous ion content by repressing iron transporters both in vitro and in vivo. In conclusion, KBs-induced mitigation of ferroptosis could represent a new therapeutic mechanism for RIPostC in treating stroke.


Subject(s)
Coenzyme A Ligases , Ferroptosis , Infarction, Middle Cerebral Artery , Ischemic Postconditioning , Ketone Bodies , Neuroprotection , Ferroptosis/physiology , Animals , Rats , Ischemic Postconditioning/methods , Ketone Bodies/metabolism , Male , Coenzyme A Ligases/metabolism , Neuroprotection/physiology , Rats, Sprague-Dawley , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Mice , Neuroprotective Agents/pharmacology , Ischemic Stroke/metabolism , Stroke/metabolism , Neurons/metabolism
18.
Brain Res Bull ; 212: 110964, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38670471

ABSTRACT

Ischemic stroke (IS), primarily caused by cerebrovascular obstruction, results in severe neurological deficits and has emerged as a leading cause of death and disability worldwide. Recently, there has been increasing exploration of the neuroprotective properties of the inert gas argon. Argon has exhibited impressive neuroprotection in many in vivo and ex vivo experiments without signs of adverse effects, coupled with the advantages of being inexpensive and easily available. However, the efficient administration strategy and underlying mechanisms of neuroprotection by argon in IS are still unclear. This review summarizes current research on the neuroprotective effects of argon in IS with the goal to provide effective guidance for argon application and to elucidate the potential mechanisms of argon neuroprotection. Early and appropriate argon administration at as high a concentration as possible offers favorable neuroprotection in IS. Argon inhalation has been shown to provide some long-term protection benefits. Argon provides the anti-oxidative stress, anti-inflammatory and anti-apoptotic cytoprotective effects mainly around Toll-like receptor 2/4 (TLR2/4), mediated by extracellular signal-regulated kinase 1/2 (ERK1/2), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), nuclear factor kappa-B (NF-ĸB) and B-cell leukemia/lymphoma 2 (Bcl-2). Therefore, argon holds significant promise as a novel clinical neuroprotective gas agent for ischemic stroke after further researches to identify the optimal application strategy and elucidate the underlying mechanism.


Subject(s)
Argon , Ischemic Stroke , Neuroprotective Agents , Argon/pharmacology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Animals , Humans , Ischemic Stroke/drug therapy , Oxidative Stress/drug effects , Neuroprotection/drug effects , Neuroprotection/physiology , Brain Ischemia/drug therapy , Brain Ischemia/metabolism
19.
Int J Mol Sci ; 25(8)2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38674030

ABSTRACT

Age-associated deep-subcortical white matter lesions (DSCLs) are an independent risk factor for dementia, displaying high levels of CD68+ microglia. This study aimed to characterize the transcriptomic profile of microglia in DSCLs and surrounding radiologically normal-appearing white matter (NAWM) compared to non-lesional control white matter. CD68+ microglia were isolated from white matter groups (n = 4 cases per group) from the Cognitive Function and Ageing Study neuropathology cohort using immuno-laser capture microdissection. Microarray gene expression profiling, but not RNA-sequencing, was found to be compatible with immuno-LCM-ed post-mortem material in the CFAS cohort and identified significantly differentially expressed genes (DEGs). Functional grouping and pathway analysis were assessed using the Database for Annotation Visualization and Integrated Discovery (DAVID) software, and immunohistochemistry was performed to validate gene expression changes at the protein level. Transcriptomic profiling of microglia in DSCLs compared to non-lesional control white matter identified 181 significant DEGs (93 upregulated and 88 downregulated). Functional clustering analysis in DAVID revealed dysregulation of haptoglobin-haemoglobin binding (Enrichment score 2.5, p = 0.017), confirmed using CD163 immunostaining, suggesting a neuroprotective microglial response to blood-brain barrier dysfunction in DSCLs. In NAWM versus control white matter, microglia exhibited 347 DEGs (209 upregulated, 138 downregulated), with significant dysregulation of protein de-ubiquitination (Enrichment score 5.14, p < 0.001), implying an inability to maintain protein homeostasis in NAWM that may contribute to lesion spread. These findings enhance understanding of microglial transcriptomic changes in ageing white matter pathology, highlighting a neuroprotective adaptation in DSCLs microglia and a potentially lesion-promoting phenotype in NAWM microglia.


Subject(s)
Aging , Blood-Brain Barrier , Microglia , Transcriptome , White Matter , Humans , Microglia/metabolism , Microglia/pathology , White Matter/metabolism , White Matter/pathology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Male , Female , Aging/genetics , Aged , Gene Expression Profiling/methods , Aged, 80 and over , Neuroprotection/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, CD/metabolism , Antigens, CD/genetics
20.
Biol Sex Differ ; 15(1): 30, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38566248

ABSTRACT

BACKGROUND: Neonatal hypoxia ischemia (HI) related brain injury is one of the major causes of learning disabilities and memory deficits in children. In both human and animal studies, female neonate brains are less susceptible to HI than male brains. Phosphorylation of the nerve growth factor receptor TrkB has been shown to provide sex-specific neuroprotection following in vivo HI in female mice in an estrogen receptor alpha (ERα)-dependent manner. However, the molecular and cellular mechanisms conferring sex-specific neonatal neuroprotection remain incompletely understood. Here, we test whether female neonatal hippocampal neurons express autonomous neuroprotective properties and assess the ability of testosterone (T) to alter this phenotype. METHODS: We cultured sexed hippocampal neurons from ERα+/+ and ERα-/- mice and subjected them to 4 h oxygen glucose deprivation and 24 h reoxygenation (4-OGD/24-REOX). Sexed hippocampal neurons were treated either with vehicle control (VC) or the TrkB agonist 7,8-dihydroxyflavone (7,8-DHF) following in vitro ischemia. End points at 24 h REOX were TrkB phosphorylation (p-TrkB) and neuronal survival assessed by immunohistochemistry. In addition, in vitro ischemia-mediated ERα gene expression in hippocampal neurons were investigated following testosterone (T) pre-treatment and TrkB antagonist therapy via q-RTPCR. Multifactorial analysis of variance was conducted to test for significant differences between experimental conditions. RESULTS: Under normoxic conditions, administration of 3 µM 7,8-DHF resulted an ERα-dependent increase in p-TrkB immunoexpression that was higher in female, as compared to male neurons. Following 4-OGD/24-REOX, p-TrkB expression increased 20% in both male and female ERα+/+ neurons. However, with 3 µM 7,8-DHF treatment p-TrkB expression increased further in female neurons by 2.81 ± 0.79-fold and was ERα dependent. 4-OGD/24-REOX resulted in a 56% increase in cell death, but only female cells were rescued with 3 µM 7,8-DHF, again in an ERα dependent manner. Following 4-OGD/3-REOX, ERα mRNA increased ~ 3 fold in female neurons. This increase was blocked with either the TrkB antagonist ANA-12 or pre-treatment with T. Pre-treatment with T also blocked the 7,8-DHF- dependent sex-specific neuronal survival in female neurons following 4-OGD/24-REOX. CONCLUSIONS: OGD/REOX results in sex-dependent TrkB phosphorylation in female neurons that increases further with 7,8-DHF treatment. TrkB phosphorylation by 7,8-DHF increased ERα mRNA expression and promoted cell survival preferentially in female hippocampal neurons. The sex-dependent neuroprotective actions of 7,8-DHF were blocked by either ANA-12 or by T pre-treatment. These results are consistent with a model for a female-specific neuroprotective pathway in hippocampal neurons in response to hypoxia. The pathway is activated by 7,8-DHF, mediated by TrkB phosphorylation, dependent on ERα and blocked by pre-exposure to T.


Subject(s)
Estrogen Receptor alpha , Neuroprotective Agents , Child , Female , Animals , Male , Mice , Humans , Estrogen Receptor alpha/metabolism , Neuroprotection , Sex Characteristics , Testosterone/pharmacology , Testosterone/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/metabolism , Neurons/metabolism , Hippocampus/metabolism , Ischemia , Hypoxia/metabolism , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL